1
|
Nakamura T, Nakamura HM, Iwasaki Y, Enomoto-Iwamoto M, Nakashima N, Fukumoto S, Pacifici M, Iwamoto M, Wakamori M. Molecular mechanism for transcriptional regulation of the parathyroid hormone gene by Epiprofin. FEBS J 2025. [PMID: 40164571 DOI: 10.1111/febs.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/29/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Abstract
Epiprofin (Epfn), an Sp/KLF family transcription factor that regulates cell proliferation and determines cell fates, is essential for normal skin, hair follicle, and tooth development. We found that Epfn was expressed in parathyroid glands, and Epfn-knockout mice displayed elevated serum parathyroid hormone (PTH) concentrations, decreased bone volume, and intracranial ectopic calcification. To investigate the role of Epfn in the regulation of PTH expression, parathyroid gland explant and parathyroid cell line culture methods were used. Epfn expression was found to be upregulated in response to an increase in extracellular calcium concentration, whereas PTH expression was downregulated, thus demonstrating an inverse correlation. Forced expression of Epfn inhibited PTH gene expression and PTH promoter reporter activity in parathyroid cells. In addition, with a high extracellular calcium concentration, Epfn silencing in cultured parathyroid glands failed to block PTH gene expression. ChIP-qPCR analysis also revealed Epfn binding in the proximal region of the PTH promoter, which was accelerated in the presence of a high concentration of calcium ions. The results from our in vitro and ex vivo analyses suggest that Epfn is a newly identified negative regulator of PTH transcription by regulating the proximal PTH promoter. Furthermore, the expression of Epfn was significantly reduced in parathyroid adenomas of primary hyperparathyroidism patients. The identification of Epfn as a potential therapeutic target for the control of PTH production in hyperparathyroidism patients opens new avenues for targeted treatment approaches.
Collapse
Affiliation(s)
- Takashi Nakamura
- Division of Molecular Pharmacology & Cell Biophysics, Department of Disease Management Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Hannah M Nakamura
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yasumasa Iwasaki
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kochi University, Japan
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Noriaki Nakashima
- Department of Breast Cancer and Endocrine Surgery, Tohoku University Hospital, Sendai, Japan
| | - Satoshi Fukumoto
- Division of Pediatric Dentistry, Kyushu University Graduate School of Dentistry, Fukuoka, Japan
| | - Maurizio Pacifici
- Division of Orthopedic Surgery, Department of Surgery, Children's Hospital of Philadelphia, PA, USA
| | - Masahiro Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Minoru Wakamori
- Division of Molecular Pharmacology & Cell Biophysics, Department of Disease Management Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
2
|
Ruolo I, Napolitano S, Postiglione L, Napolitano G, Ballabio A, di Bernardo D. Investigation of dynamic regulation of TFEB nuclear shuttling by microfluidics and quantitative modelling. Commun Biol 2025; 8:443. [PMID: 40089585 PMCID: PMC11910602 DOI: 10.1038/s42003-025-07870-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Transcription Factor EB (TFEB) controls lysosomal biogenesis and autophagy in response to nutritional status and other stress factors. Although its regulation by nuclear translocation is known to involve a complex network of well-studied regulatory processes, the precise contribution of each of these mechanisms is unclear. Using microfluidics technology and real-time imaging coupled with mathematical modelling, we explored the dynamic regulation of TFEB under different conditions. We found that TFEB nuclear translocation upon nutrient deprivation happens in two phases: a fast one characterised by a transient boost in TFEB dephosphorylation dependent on transient calcium release mediated by mucolipin 1 (MCOLN1) followed by activation of the Calcineurin phosphatase, and a slower one driven by inhibition of mTORC1-dependent phosphorylation of TFEB. Upon refeeding, TFEB cytoplasmic relocalisation kinetics are determined by Exportin 1 (XPO1). Collectively, our results show how different mechanisms interact to regulate TFEB activation and the power of microfluidics and quantitative modelling to elucidate complex biological mechanisms.
Collapse
Affiliation(s)
- Iacopo Ruolo
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sara Napolitano
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Institut Pasteur, Inria, Université Paris Cité, Paris, France
| | - Lorena Postiglione
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Gennaro Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
- SSM School for Advanced Studies, Federico II University, Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, US
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, US
| | - Diego di Bernardo
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy.
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.
- SSM School for Advanced Studies, Federico II University, Naples, Italy.
| |
Collapse
|
3
|
Young M, Booth DM, Smith D, Tigano M, Hajnόczky G, Joseph SK. Transcriptional regulation in the absence of inositol trisphosphate receptor calcium signaling. Front Cell Dev Biol 2024; 12:1473210. [PMID: 39712573 PMCID: PMC11659226 DOI: 10.3389/fcell.2024.1473210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The activation of IP3 receptor (IP3R) Ca2+ channels generates agonist-mediated Ca2+ signals that are critical for the regulation of a wide range of biological processes. It is therefore surprising that CRISPR induced loss of all three IP3R isoforms (TKO) in HEK293 and HeLa cell lines yields cells that can survive, grow and divide, albeit more slowly than wild-type cells. In an effort to understand the adaptive mechanisms involved, we have examined the activity of key Ca2+ dependent transcription factors (NFAT, CREB and AP-1) and signaling pathways using luciferase-reporter assays, phosphoprotein immunoblots and whole genome transcriptomic studies. In addition, the diacylglycerol arm of the signaling pathway was investigated with protein kinase C (PKC) inhibitors and siRNA knockdown. The data showed that agonist-mediated NFAT activation was lost but CREB activation was maintained in IP3R TKO cells. Under base-line conditions transcriptome analysis indicated the differential expression of 828 and 311 genes in IP3R TKO HEK293 or HeLa cells, respectively, with only 18 genes being in common. Three main adaptations in TKO cells were identified in this study: 1) increased basal activity of NFAT, CREB and AP-1; 2) an increased reliance on Ca2+- insensitive PKC isoforms; and 3) increased production of reactive oxygen species and upregulation of antioxidant defense enzymes. We suggest that whereas wild-type cells rely on a Ca2+ and DAG signal to respond to stimuli, the TKO cells utilize the adaptations to allow key signaling pathways (e.g., PKC, Ras/MAPK, CREB) to transition to the activated state using a DAG signal alone.
Collapse
Affiliation(s)
- Michael Young
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - David M. Booth
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - David Smith
- Center for Single Cell Biology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Marco Tigano
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Gyӧrgy Hajnόczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Suresh K. Joseph
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
4
|
Zheng B, Zhang X, Kong X, Li J, Huang B, Li H, Ji Z, Wei X, Tao S, Shan Z, Ling Z, Liu J, Chen J, Zhao F. S1P regulates intervertebral disc aging by mediating endoplasmic reticulum-mitochondrial calcium ion homeostasis. JCI Insight 2024; 9:e177789. [PMID: 39316443 PMCID: PMC11601718 DOI: 10.1172/jci.insight.177789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 09/18/2024] [Indexed: 09/26/2024] Open
Abstract
As the aging process progresses, age-related intervertebral disc degeneration (IVDD) is becoming an emerging public health issue. Site-1 protease (S1P) has recently been found to be associated with abnormal spinal development in patients with mutations and has multiple biological functions. Here, we discovered a reduction of S1P in degenerated and aging intervertebral discs, primarily regulated by DNA methylation. Furthermore, through drug treatment and siRNA-mediated S1P knockdown, nucleus pulposus cells were more prone to exhibit degenerative and aging phenotypes. Conditional KO of S1P in mice resulted in spinal developmental abnormalities and premature aging. Mechanistically, S1P deficiency impeded COP II-mediated transport vesicle formation, which leads to protein retention in the endoplasmic reticulum (ER) and subsequently ER distension. ER distension increased the contact between the ER and mitochondria, disrupting ER-to-mitochondria calcium flow and resulting in mitochondrial dysfunction and energy metabolism disturbance. Finally, using 2-APB to inhibit calcium ion channels and the senolytic drug dasatinib and quercetin (D + Q) partially rescued the aging and degenerative phenotypes caused by S1P deficiency. In conclusion, our findings suggest that S1P is a critical factor in causing IVDD in the process of aging and highlight the potential of targeting S1P as a therapeutic approach for age-related IVDD.
Collapse
Affiliation(s)
- Bingjie Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xuyang Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiangxi Kong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jie Li
- Department of Orthopaedic Surgery, Ningbo Medical Center Li Huili Hospital, Ningbo, China
| | - Bao Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hui Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhongyin Ji
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiaoan Wei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Siyue Tao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhi Shan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zemin Ling
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Junhui Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jian Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Mackiewicz J, Tomczak J, Lisek M, Sakowicz A, Guo F, Boczek T. NFATc4 Knockout Promotes Neuroprotection and Retinal Ganglion Cell Regeneration After Optic Nerve Injury. Mol Neurobiol 2024; 61:9383-9401. [PMID: 38639863 PMCID: PMC11496353 DOI: 10.1007/s12035-024-04129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/19/2024] [Indexed: 04/20/2024]
Abstract
Retinal ganglion cells (RGCs), neurons transmitting visual information via the optic nerve, fail to regenerate their axons after injury. The progressive loss of RGC function underlies the pathophysiology of glaucoma and other optic neuropathies, often leading to irreversible blindness. Therefore, there is an urgent need to identify the regulators of RGC survival and the regenerative program. In this study, we investigated the role of the family of transcription factors known as nuclear factor of activated T cells (NFAT), which are expressed in the retina; however, their role in RGC survival after injury is unknown. Using the optic nerve crush (ONC) model, widely employed to study optic neuropathies and central nervous system axon injury, we found that NFATc4 is specifically but transiently up-regulated in response to mechanical injury. In the injured retina, NFATc4 immunolocalized primarily to the ganglionic cell layer. Utilizing NFATc4-/- and NFATc3-/- mice, we demonstrated that NFATc4, but not NFATc3, knockout increased RGC survival, improved retina function, and delayed axonal degeneration. Microarray screening data, along with decreased immunostaining of cleaved caspase-3, revealed that NFATc4 knockout was protective against ONC-induced degeneration by suppressing pro-apoptotic signaling. Finally, we used lentiviral-mediated NFATc4 delivery to the retina of NFATc4-/- mice and reversed the pro-survival effect of NFATc4 knockout, conclusively linking the enhanced survival of injured RGCs to NFATc4-dependent mechanisms. In summary, this study is the first to demonstrate that NFATc4 knockout may confer transient RGC neuroprotection and decelerate axonal degeneration after injury, providing a potent therapeutic strategy for optic neuropathies.
Collapse
Affiliation(s)
- Joanna Mackiewicz
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Julia Tomczak
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Malwina Lisek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Lodz, Poland
| | - Feng Guo
- Department of Pharmaceutical Toxicology, China Medical University, Shenyang, China.
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
6
|
Ceci M, Bonvissuto D, Papetti F, Silvestri F, Sette C, Catalani E, Cervia D, Gornati R, Romano N. RACK1 contributes to the upregulation of embryonic genes in a model of cardiac hypertrophy. Sci Rep 2024; 14:25698. [PMID: 39465301 PMCID: PMC11514175 DOI: 10.1038/s41598-024-76138-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024] Open
Abstract
Receptors for activated C kinases (RACKs) have been shown to coordinate PKC-mediated hypertrophic signalling in mice. However, little information is available on its participation in embryonic gene expression. This study investigated the involvement of RACK1 in the expression of embryonic genes in a zebrafish (ZF) ex vivo heart culture model by using phenylephrine (PE) or a growth factors cocktail (GFs) as a prohypertrophic/regeneration stimulus. Blebbistatin (BL) inhibition has also been studied for its ability to block the signal transduction actions of some PEs. qRT‒PCR and immunoblot analyses confirmed the upregulation of RACK1 in the PE- and GFs-treated groups. BL administration counteracted PE-induced hypertrophy and downregulated RACK1 expression. Immunohistochemical analyses of the heart revealed the colocalization of RACK1 and embryonic genes, namely, Gata4, Wt1, and Nfat2, under stimulation, whereas these genes were expressed at lower levels in the BL treatment group. Culturing ZF heart cells activated via GFs treatment increased the expression of RACK1. The overexpression of RACK1 induced by the transfection of recombinant RACK1 cDNA in ZF heart cells increased the expression of embryonic genes, especially after one week of GFs treatment. In summary, these results support the involvement of RACK1 in the induction of embryonic genes during cardiac hypertrophy/GFs stimulation in a fish heart model, which can be used as an alternative study model for mammals.
Collapse
Affiliation(s)
| | | | | | | | - Claudio Sette
- DNHA, Catholic University of Sacred Heart, Rome, Italy
- IRCCS, Policlinico A. Gemelli Foundation, Rome, Italy
| | | | | | | | | |
Collapse
|
7
|
Katsuyama E, Humbel M, Suarez-Fueyo A, Satyam A, Yoshida N, Kyttaris VC, Tsokos MG, Tsokos GC. CD38 in SLE CD4 T cells promotes Ca 2+ flux and suppresses interleukin-2 production by enhancing the expression of GM2 on the surface membrane. Nat Commun 2024; 15:8304. [PMID: 39333474 PMCID: PMC11436706 DOI: 10.1038/s41467-024-52617-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 09/17/2024] [Indexed: 09/29/2024] Open
Abstract
CD38 has emerged as a potential therapeutic target for patients with systemic lupus erythematosus (SLE) but it is not known whether CD38 alters CD4+ T cell function. Using primary human T cells and CD38-sufficient and CD38-deficient Jurkat T cells, we demonstrate that CD38 shifts the T cell lipid profile of gangliosides from GM3 to GM2 by upregulating B4GALNT1 in a Sirtuin 1-dependent manner. Enhanced expression of GM2 causes ER stress by enhancing Ca2+ flux through the PLCγ1-IP3 pathway. Interestingly, correction of the calcium overload by an IP3 receptor inhibitor, but not by a store-operated calcium entry (SOCE) inhibitor, improves IL-2 production by CD4+ T cells in SLE. This study demonstrates that CD38 affects calcium homeostasis in CD4+ T cells by controlling cell membrane lipid composition that results in suppressed IL-2 production. CD38 inhibition with biologics or small drugs should be expected to benefit patients with SLE.
Collapse
Affiliation(s)
- Eri Katsuyama
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Morgane Humbel
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Abel Suarez-Fueyo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Abhigyan Satyam
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Nobuya Yoshida
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Vasileios C Kyttaris
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| |
Collapse
|
8
|
Shi Q, Sun X, Zhang H, Yang L, Fu Y, Wang G, Su Y, Li W, Li W. PLC-CN-NFAT1 signaling-mediated Aβ and IL-1β crosstalk synergistically promotes hippocampal neuronal damage. Int Immunopharmacol 2024; 134:112259. [PMID: 38749336 DOI: 10.1016/j.intimp.2024.112259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/18/2024] [Accepted: 05/10/2024] [Indexed: 06/03/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. Neuronal calcium overload plays an important role in Aβ deposition and neuroinflammation, which are strongly associated with AD. However, the specific mechanisms by which calcium overload contributes to neuroinflammation and AD and the relationship between them have not been elucidated. Phospholipase C (PLC) is involved in regulation of calcium homeostasis, and CN-NFAT1 signaling is dependent on intracellular Ca2+ ([Ca2+]i) to regulate transcription of genes. Therefore, we hypothesized that the PLC-CN-NFAT1 signaling might mediate the interaction between Aβ and inflammation to promote neuronal injury in AD. In this experiment, the results showed that the levels of Aβ, IL-1β and [Ca2+]i in the hippocampal primary neurons of APP/PS1 mice (APP neurons) were significantly increased. IL-1β exposure also significantly increased Aβ and [Ca2+]i in HT22 cells, suggesting a close association between Aβ and IL-1β in the development of AD. Furthermore, PLC activation induced significant calcium homeostasis imbalance, cell apoptosis, Aβ and ROS production, and significantly increased expressions of CN and NFAT1, while PLC inhibitor significantly reversed these changes in APP neurons and IL-1β-induced HT22 cells. Further results indicated that PLC activation significantly increased the expressions of NOX2, APP, BACE1, and NCSTN, which were inhibited by PLC inhibitor in APP neurons and IL-1β-induced HT22 cells. All indications point to a synergistic interaction between Aβ and IL-1β by activating the PLC-CN-NFAT1 signal, ultimately causing a vicious cycle, resulting in neuronal damage in AD. The study may provide a new idea and target for treatment of AD.
Collapse
Affiliation(s)
- Qifeng Shi
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Xiangyu Sun
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Hui Zhang
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Liu Yang
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yinglin Fu
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Guohang Wang
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yong Su
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Weiping Li
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
9
|
Diercks BP. The importance of Ca 2+ microdomains for the adaptive immune response. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119710. [PMID: 38522726 DOI: 10.1016/j.bbamcr.2024.119710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 03/26/2024]
Abstract
Calcium signaling stands out as the most widespread and universally used signaling system and is of utmost importance for immunity. Controlled elevations in cytosolic and organellar Ca2+ concentrations in T cells control complex and essential effector functions including proliferation, differentiation, cytokine secretion, and cytotoxicity, among others. Additionally, disruptions in Ca2+ regulation in T cells contribute to diverse autoimmune, inflammatory, and immunodeficiency conditions. Among the initial intracellular signals, which occurring even before T cell receptor (TCR) stimulation are highly localized, spatially and temporally restricted so-called Ca2+ microdomains, caused by adhesion to extracellular matrix proteins (ECM proteins). The Ca2+ microdomains present both before and within the initial seconds following TCR stimulation are likely to play a crucial role in fine-tuning the downstream activity of T cell activation and thus, shaping an adaptive immune response. In this review, the emphasis is on the recent advances of adhesion-dependent Ca2+ microdomains (ADCM) in the absence of TCR stimulation, initial Ca2+ microdomains evoked by TCR stimulation (TDCM), the downstream signaling processes as well as possible therapeutic targets for interventions.
Collapse
Affiliation(s)
- Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
10
|
Brock VJ, Lory NC, Möckl F, Birus M, Stähler T, Woelk LM, Jaeckstein M, Heeren J, Koch-Nolte F, Rissiek B, Mittrücker HW, Guse AH, Werner R, Diercks BP. Time-resolved role of P2X4 and P2X7 during CD8 + T cell activation. Front Immunol 2024; 15:1258119. [PMID: 38426095 PMCID: PMC10902106 DOI: 10.3389/fimmu.2024.1258119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/11/2024] [Indexed: 03/02/2024] Open
Abstract
CD8+ T cells are a crucial part of the adaptive immune system, responsible for combating intracellular pathogens and tumor cells. The initial activation of T cells involves the formation of highly dynamic Ca2+ microdomains. Recently, purinergic signaling was shown to be involved in the formation of the initial Ca2+ microdomains in CD4+ T cells. In this study, the role of purinergic cation channels, particularly P2X4 and P2X7, in CD8+ T cell signaling from initial events to downstream responses was investigated, focusing on various aspects of T cell activation, including Ca2+ microdomains, global Ca2+ responses, NFAT-1 translocation, cytokine expression, and proliferation. While Ca2+ microdomain formation was significantly reduced in the first milliseconds to seconds in CD8+ T cells lacking P2X4 and P2X7 channels, global Ca2+ responses over minutes were comparable between wild-type (WT) and knockout cells. However, the onset velocity was reduced in P2X4-deficient cells, and P2X4, as well as P2X7-deficient cells, exhibited a delayed response to reach a certain level of free cytosolic Ca2+ concentration ([Ca2+]i). NFAT-1 translocation, a crucial transcription factor in T cell activation, was also impaired in CD8+ T cells lacking P2X4 and P2X7. In addition, the expression of IFN-γ, a major pro-inflammatory cytokine produced by activated CD8+ T cells, and Nur77, a negative regulator of T cell activation, was significantly reduced 18h post-stimulation in the knockout cells. In line, the proliferation of T cells after 3 days was also impaired in the absence of P2X4 and P2X7 channels. In summary, the study demonstrates that purinergic signaling through P2X4 and P2X7 enhances initial Ca2+ events during CD8+ T cell activation and plays a crucial role in regulating downstream responses, including NFAT-1 translocation, cytokine expression, and proliferation on multiple timescales. These findings suggest that targeting purinergic signaling pathways may offer potential therapeutic interventions.
Collapse
Affiliation(s)
- Valerie J. Brock
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Niels Christian Lory
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Möckl
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Melina Birus
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Stähler
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Lena-Marie Woelk
- Department of Applied Medical Informatics, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department of Computational Neuroscience, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Michelle Jaeckstein
- Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H. Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - René Werner
- Department of Applied Medical Informatics, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department of Computational Neuroscience, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Pane R, Laib L, Formoso K, Détrait M, Sainte-Marie Y, Bourgailh F, Ruffenach N, Faugeras H, Simon I, Lhuillier E, Lezoualc'h F, Conte C. Macromolecular Complex Including MLL3, Carabin and Calcineurin Regulates Cardiac Remodeling. Circ Res 2024; 134:100-113. [PMID: 38084599 DOI: 10.1161/circresaha.123.323458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Cardiac hypertrophy is an intermediate stage in the development of heart failure. The structural and functional processes occurring in cardiac hypertrophy include extensive gene reprogramming, which is dependent on epigenetic regulation and chromatin remodeling. However, the chromatin remodelers and their regulatory functions involved in the pathogenesis of cardiac hypertrophy are not well characterized. METHODS Protein interaction was determined by immunoprecipitation assay in primary cardiomyocytes and mouse cardiac samples subjected or not to transverse aortic constriction for 1 week. Chromatin immunoprecipitation and DNA sequencing (ChIP-seq) experiments were performed on the chromatin of adult mouse cardiomyocytes. RESULTS We report that the calcium-activated protein phosphatase CaN (calcineurin), its endogenous inhibitory protein carabin, the STK24 (STE20-like protein kinase 3), and the histone monomethyltransferase, MLL3 (mixed lineage leukemia 3) form altogether a macromolecular complex at the chromatin of cardiomyocytes. Under basal conditions, carabin prevents CaN activation while the serine/threonine kinase STK24 maintains MLL3 inactive via phosphorylation. After 1 week of transverse aortic constriction, both carabin and STK24 are released from the CaN-MLL3 complex leading to the activation of CaN, dephosphorylation of MLL3, and in turn, histone H3 lysine 4 monomethylation. Selective cardiac MLL3 knockdown mitigates hypertrophy, and chromatin immunoprecipitation and DNA sequencing analysis demonstrates that MLL3 is de novo recruited at the transcriptional start site of genes implicated in cardiomyopathy in stress conditions. We also show that CaN and MLL3 colocalize at chromatin and that CaN activates MLL3 histone methyl transferase activity at distal intergenic regions under hypertrophic conditions. CONCLUSIONS Our study reveals an unsuspected epigenetic mechanism of CaN that directly regulates MLL3 histone methyl transferase activity to promote cardiac remodeling.
Collapse
Affiliation(s)
- Roberto Pane
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Loubna Laib
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Karina Formoso
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Maximin Détrait
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Yannis Sainte-Marie
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Florence Bourgailh
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Nolan Ruffenach
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Hanamée Faugeras
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Ilias Simon
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Emeline Lhuillier
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
- GeT-Sante, Plateforme Genome et Transcriptome, GenoToul, Toulouse, France (E.L.)
| | - Frank Lezoualc'h
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| | - Caroline Conte
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université de Toulouse III-Paul Sabatier, France (R.P., L.L., K.F., M.D.., Y.S.-M., F.B., N.R., H.F., I.S., E.L., F.L., C.C.)
| |
Collapse
|
12
|
Gross S, Womer L, Kappes DJ, Soboloff J. Multifaceted control of T cell differentiation by STIM1. Trends Biochem Sci 2023; 48:1083-1097. [PMID: 37696713 PMCID: PMC10787584 DOI: 10.1016/j.tibs.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023]
Abstract
In T cells, stromal interaction molecule (STIM) and Orai are dispensable for conventional T cell development, but critical for activation and differentiation. This review focuses on novel STIM-dependent mechanisms for control of Ca2+ signals during T cell activation and its impact on mitochondrial function and transcriptional activation for control of T cell differentiation and function. We highlight areas that require further work including the roles of plasma membrane Ca2+ ATPase (PMCA) and partner of STIM1 (POST) in controlling Orai function. A major knowledge gap also exists regarding the independence of T cell development from STIM and Orai, despite compelling evidence that it requires Ca2+ signals. Resolving these and other outstanding questions ensures that the field will remain active for many years to come.
Collapse
Affiliation(s)
- Scott Gross
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Lauren Womer
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | | | - Jonathan Soboloff
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Department of Cancer and Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
13
|
Kolbe MR, Hohmann T, Hohmann U, Maronde E, Golbik R, Prell J, Illert J, Strauss C, Dehghani F. Elucidation of GPR55-Associated Signaling behind THC and LPI Reducing Effects on Ki67-Immunoreactive Nuclei in Patient-Derived Glioblastoma Cells. Cells 2023; 12:2646. [PMID: 37998380 PMCID: PMC10670585 DOI: 10.3390/cells12222646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
GPR55 is involved in many physiological and pathological processes. In cancer, GPR55 has been described to show accelerating and decelerating effects in tumor progression resulting from distinct intracellular signaling pathways. GPR55 becomes activated by LPI and various plant-derived, endogenous, and synthetic cannabinoids. Cannabinoids such as THC exerted antitumor effects by inhibiting tumor cell proliferation or inducing apoptosis. Besides its effects through CB1 and CB2 receptors, THC modulates cellular responses among others via GPR55. Previously, we reported a reduction in Ki67-immunoreactive nuclei of human glioblastoma cells after GPR55 activation in general by THC and in particular by LPI. In the present study, we investigated intracellular mechanisms leading to an altered number of Ki67+ nuclei after stimulation of GPR55 by LPI and THC. Pharmacological analyses revealed a strongly involved PLC-IP3 signaling and cell-type-specific differences in Gα-, Gβγ-, RhoA-ROCK, and calcineurin signaling. Furthermore, immunochemical visualization of the calcineurin-dependent transcription factor NFAT revealed an unchanged subcellular localization after THC or LPI treatment. The data underline the cell-type-specific diversity of GPR55-associated signaling pathways in coupling to intracellular G proteins. Furthermore, this diversity might determine the outcome and the individual responsiveness of tumor cells to GPR55 stimulation by cannabin oids.
Collapse
Affiliation(s)
- Marc Richard Kolbe
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle (Saale), Germany; (M.R.K.); (T.H.); (U.H.)
| | - Tim Hohmann
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle (Saale), Germany; (M.R.K.); (T.H.); (U.H.)
| | - Urszula Hohmann
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle (Saale), Germany; (M.R.K.); (T.H.); (U.H.)
| | - Erik Maronde
- Department of Anatomy II, Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| | - Ralph Golbik
- Charles Tanford Protein Centre, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120 Halle (Saale), Germany;
| | - Julian Prell
- Department of Neurosurgery, Medical Faculty, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany; (J.P.); (J.I.); (C.S.)
| | - Jörg Illert
- Department of Neurosurgery, Medical Faculty, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany; (J.P.); (J.I.); (C.S.)
| | - Christian Strauss
- Department of Neurosurgery, Medical Faculty, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany; (J.P.); (J.I.); (C.S.)
| | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle (Saale), Germany; (M.R.K.); (T.H.); (U.H.)
| |
Collapse
|
14
|
Nieto-Felipe J, Macias-Diaz A, Sanchez-Collado J, Berna-Erro A, Jardin I, Salido GM, Lopez JJ, Rosado JA. Role of Orai-family channels in the activation and regulation of transcriptional activity. J Cell Physiol 2023; 238:714-726. [PMID: 36952615 DOI: 10.1002/jcp.30971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/12/2023] [Accepted: 01/27/2023] [Indexed: 03/25/2023]
Abstract
Store operated Ca2+ entry (SOCE) is a cornerstone for the maintenance of intracellular Ca2+ homeostasis and the regulation of a variety of cellular functions. SOCE is mediated by STIM and Orai proteins following the activation of inositol 1,4,5-trisphosphate receptors. Then, a reduction of the endoplasmic reticulum intraluminal Ca2+ concentration is sensed by STIM proteins, which undergo a conformational change and activate plasma membrane Ca2+ channels comprised by Orai proteins. STIM1/Orai-mediated Ca2+ signals are finely regulated and modulate the activity of different transcription factors, including certain isoforms of the nuclear factor of activated T-cells, the cAMP-response element binding protein, the nuclear factor κ-light chain-enhancer of activated B cells, c-fos, and c-myc. These transcription factors associate SOCE with a plethora of signaling events and cellular functions. Here we provide an overview of the current knowledge about the role of Orai channels in the regulation of transcription factors through Ca2+ -dependent signaling pathways.
Collapse
Affiliation(s)
- Joel Nieto-Felipe
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Alvaro Macias-Diaz
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Jose Sanchez-Collado
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Alejandro Berna-Erro
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Isaac Jardin
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Gines M Salido
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Jose J Lopez
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Juan A Rosado
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| |
Collapse
|
15
|
Liu X, Chang Y, Choi S, Cai C, Zhang X, Pan Z. Blocking Store-Operated Ca 2+ Entry to Protect HL-1 Cardiomyocytes from Epirubicin-Induced Cardiotoxicity. Cells 2023; 12:723. [PMID: 36899859 PMCID: PMC10000558 DOI: 10.3390/cells12050723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Epirubicin (EPI) is one of the most widely used anthracycline chemotherapy drugs, yet its cardiotoxicity severely limits its clinical application. Altered intracellular Ca2+ homeostasis has been shown to contribute to EPI-induced cell death and hypertrophy in the heart. While store-operated Ca2+ entry (SOCE) has recently been linked with cardiac hypertrophy and heart failure, its role in EPI-induced cardiotoxicity remains unknown. Using a publicly available RNA-seq dataset of human iPSC-derived cardiomyocytes, gene analysis showed that cells treated with 2 µM EPI for 48 h had significantly reduced expression of SOCE machinery genes, e.g., Orai1, Orai3, TRPC3, TRPC4, Stim1, and Stim2. Using HL-1, a cardiomyocyte cell line derived from adult mouse atria, and Fura-2, a ratiometric Ca2+ fluorescent dye, this study confirmed that SOCE was indeed significantly reduced in HL-1 cells treated with EPI for 6 h or longer. However, HL-1 cells presented increased SOCE as well as increased reactive oxygen species (ROS) production at 30 min after EPI treatment. EPI-induced apoptosis was evidenced by disruption of F-actin and increased cleavage of caspase-3 protein. The HL-1 cells that survived to 24 h after EPI treatment demonstrated enlarged cell sizes, up-regulated expression of brain natriuretic peptide (a hypertrophy marker), and increased NFAT4 nuclear translocation. Treatment by BTP2, a known SOCE blocker, decreased the initial EPI-enhanced SOCE, rescued HL-1 cells from EPI-induced apoptosis, and reduced NFAT4 nuclear translocation and hypertrophy. This study suggests that EPI may affect SOCE in two phases: the initial enhancement phase and the following cell compensatory reduction phase. Administration of a SOCE blocker at the initial enhancement phase may protect cardiomyocytes from EPI-induced toxicity and hypertrophy.
Collapse
Affiliation(s)
- Xian Liu
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
- Department of Graduate Nursing, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Yan Chang
- Department of Graduate Nursing, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
- Bone and Muscle Research Center, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Sangyong Choi
- Department of Graduate Nursing, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Chuanxi Cai
- Department of Surgery, Division of Surgical Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Xiaoli Zhang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Zui Pan
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
- Department of Graduate Nursing, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
- Bone and Muscle Research Center, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| |
Collapse
|
16
|
Saghatelyan A. Calcium signaling as an integrator and decoder of niche factors to control somatic stem cell quiescence and activation. FEBS J 2023; 290:677-683. [PMID: 34797958 DOI: 10.1111/febs.16289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/26/2021] [Accepted: 11/18/2021] [Indexed: 02/04/2023]
Abstract
Somatic stem cells (SSCs) play a major role in tissue homeostasis and respond to a panoply of micro-environmental cues by adjusting their quiescence and activation profiles. How these cells integrate and decode multiple niche signals remains elusive. In recent years, Ca2+ signaling has emerged as one of the key intracellular pathways that allow stem cells to dynamically adjust their fate and either to remain quiescent for future needs or to become activated to generate new progeny. Interestingly, not only distinct Ca2+ signatures are associated with the quiescence and activation states of stem cells, but also various extracellular cues impinge on Ca2+ pathways to dynamically regulate the responses of stem cells to different niche signals. This Viewpoint article deals with how Ca2+ signaling may be used to decode and integrate different niche factors and how Ca2+ fluctuations of distinct amplitudes, frequencies, and overall intracellular levels may trigger the differential gene transcription program. Knowledge about mechanisms that allow SSCs to translate the complexity of extracellular niche signaling into intrinsic states of cell quiescence and activation is crucial for understanding life-long tissue homeostasis and regeneration.
Collapse
Affiliation(s)
- Armen Saghatelyan
- CERVO Brain Research Center, Quebec City, Quebec, Canada.,Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
17
|
Parekh AB, Parekh LBC. "Study the Past if You Would Define the Future."-Confucius. FUNCTION (OXFORD, ENGLAND) 2023; 4:zqac073. [PMID: 36686642 PMCID: PMC9850269 DOI: 10.1093/function/zqac073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023]
|
18
|
Macrophage NFATC2 mediates angiogenic signaling during mycobacterial infection. Cell Rep 2022; 41:111817. [PMID: 36516756 PMCID: PMC9880963 DOI: 10.1016/j.celrep.2022.111817] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
During mycobacterial infections, pathogenic mycobacteria manipulate both host immune and stromal cells to establish and maintain a productive infection. In humans, non-human primates, and zebrafish models of infection, pathogenic mycobacteria produce and modify the specialized lipid trehalose 6,6'-dimycolate (TDM) in the bacterial cell envelope to drive host angiogenesis toward the site of forming granulomas, leading to enhanced bacterial growth. Here, we use the zebrafish-Mycobacterium marinum infection model to define the signaling basis of the host angiogenic response. Through intravital imaging and cell-restricted peptide-mediated inhibition, we identify macrophage-specific activation of NFAT signaling as essential to TDM-mediated angiogenesis in vivo. Exposure of cultured human cells to Mycobacterium tuberculosis results in robust induction of VEGFA, which is dependent on a signaling pathway downstream of host TDM detection and culminates in NFATC2 activation. As granuloma-associated angiogenesis is known to serve bacterial-beneficial roles, these findings identify potential host targets to improve tuberculosis disease outcomes.
Collapse
|
19
|
Curcic S, Erkan-Candag H, Pilic J, Malli R, Wiedner P, Tiapko O, Groschner K. TRPC3 governs the spatiotemporal organization of cellular Ca 2+ signatures by functional coupling to IP 3 receptors. Cell Calcium 2022; 108:102670. [PMID: 36375273 DOI: 10.1016/j.ceca.2022.102670] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/12/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Communication between TRPC channels and IP3 receptors (IP3R) is considered pivotal in the generation of spatiotemporal Ca2+signaling patterns. Here we revisited the role of TRPC3-IP3R coupling for local Ca2+ signaling within TRPC3-harbouring micro/nanodomains using R-GECO as a reporter, fused to the channel´s C-terminus. Cytoplasmic Ca2+ changes at TRPC3 originated from both the entry of Ca2+ through the TRPC channel and Ca2+ mobilization via IP3R. Local Ca2+ changes at TRPC3 channels expressed in HEK293 cells were predominantly biphasic with IP3R-dependent initial Ca2+ transients, while exclusively monophasic signals were recorded when all three IP3R isoforms were lacking. Abrogation of Ca2+ entry through TRPC3 by point mutations, which impair Ca2+ permeability (E630Q), cation permeation (E630K), or DAG sensitivity (G652A), promoted microdomain Ca2+ oscillations. Ca2+ signals at E630Q, E630K, and G652A channels featured initial Ca2+ transients along with oscillatory activity. Similarly, when extracellular Ca2+ was omitted, IP3R-mediated Ca2+ transients and Ca2+ oscillations were promoted at the cytoplasmic face of wild-type TRPC3 channels. By contrast, oscillations, as well as initial Ca2+ transients, were virtually lacking, when the TRPC3 channels were sensitized by preexposure to low-level PLC activity. TIRF imaging provided evidence for dynamic colocalization of TRPC3 and IP3R. We suggest that TRPC3-mediated Ca2+ entry controls IP3R activity at ER-PM junctions to determine Ca2+ signaling signatures and enable specificity of downstream signaling.
Collapse
Affiliation(s)
- Sanja Curcic
- Gottfried-Schatz Research Center (Biophysics), Medical University of Graz, Austria.
| | - Hazel Erkan-Candag
- Gottfried-Schatz Research Center (Biophysics), Medical University of Graz, Austria
| | - Johannes Pilic
- Gottfried-Schatz Research Center (Molecular Biology and Biochemistry), Medical University of Graz, Austria
| | - Roland Malli
- Gottfried-Schatz Research Center (Molecular Biology and Biochemistry), Medical University of Graz, Austria
| | - Patrick Wiedner
- Gottfried-Schatz Research Center (Biophysics), Medical University of Graz, Austria
| | - Oleksandra Tiapko
- Gottfried-Schatz Research Center (Biophysics), Medical University of Graz, Austria
| | - Klaus Groschner
- Gottfried-Schatz Research Center (Biophysics), Medical University of Graz, Austria.
| |
Collapse
|
20
|
Jardin I, Berna-Erro A, Nieto-Felipe J, Macias A, Sanchez-Collado J, Lopez JJ, Salido GM, Rosado JA. Similarities and Differences between the Orai1 Variants: Orai1α and Orai1β. Int J Mol Sci 2022; 23:ijms232314568. [PMID: 36498894 PMCID: PMC9735889 DOI: 10.3390/ijms232314568] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Orai1, the first identified member of the Orai protein family, is ubiquitously expressed in the animal kingdom. Orai1 was initially characterized as the channel responsible for the store-operated calcium entry (SOCE), a major mechanism that allows cytosolic calcium concentration increments upon receptor-mediated IP3 generation, which results in intracellular Ca2+ store depletion. Furthermore, current evidence supports that abnormal Orai1 expression or function underlies several disorders. Orai1 is, together with STIM1, the key element of SOCE, conducting the Ca2+ release-activated Ca2+ (CRAC) current and, in association with TRPC1, the store-operated Ca2+ (SOC) current. Additionally, Orai1 is involved in non-capacitative pathways, as the arachidonate-regulated or LTC4-regulated Ca2+ channel (ARC/LRC), store-independent Ca2+ influx activated by the secretory pathway Ca2+-ATPase (SPCA2) and the small conductance Ca2+-activated K+ channel 3 (SK3). Furthermore, Orai1 possesses two variants, Orai1α and Orai1β, the latter lacking 63 amino acids in the N-terminus as compared to the full-length Orai1α form, which confers distinct features to each variant. Here, we review the current knowledge about the differences between Orai1α and Orai1β, the implications of the Ca2+ signals triggered by each variant, and their downstream modulatory effect within the cell.
Collapse
|
21
|
Nuanced Interactions between AKAP79 and STIM1 with Orai1 Ca 2+ Channels at Endoplasmic Reticulum-Plasma Membrane Junctions Sustain NFAT Activation. Mol Cell Biol 2022; 42:e0017522. [PMID: 36317924 PMCID: PMC9670898 DOI: 10.1128/mcb.00175-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A-kinase anchoring protein 79 (AKAP79) is a human scaffolding protein that organizes Ca2+/calmodulin-dependent protein phosphatase calcineurin, calmodulin, cAMP-dependent protein kinase, protein kinase C, and the transcription factor nuclear factor of activated T cells (NFAT1) into a signalosome at the plasma membrane. Upon Ca2+ store depletion, AKAP79 interacts with the N-terminus of STIM1-gated Orai1 Ca2+ channels, enabling Ca2+ nanodomains to stimulate calcineurin. Calcineurin then dephosphorylates and activates NFAT1, which then translocates to the nucleus. A fundamental question is how signalosomes maintain long-term signaling when key effectors are released and therefore removed beyond the reach of the activating signal. Here, we show that the AKAP79-Orai1 interaction is considerably more transient than that of STIM1-Orai1. Free AKAP79, with calcineurin and NFAT1 in tow, is able to replace rapidly AKAP79 devoid of NFAT1 on Orai1, in the presence of continuous Ca2+ entry. We also show that Ca2+ nanodomains near Orai1 channels activate almost the entire cytosolic pool of NFAT1. Recycling of inactive NFAT1 from the cytoplasm to AKAP79 in the plasma membrane, coupled with the relatively weak interaction between AKAP79 and Orai1, maintain excitation-transcription coupling. By measuring rates for AKAP79-NFAT interaction, we formulate a mathematical model that simulates NFAT dynamics at the plasma membrane.
Collapse
|
22
|
Barak P, Kaur S, Scappini E, Tucker CJ, Parekh AB. Plasma Membrane Ca 2+ ATPase Activity Enables Sustained Store-operated Ca 2+ Entry in the Absence of a Bulk Cytosolic Ca 2+ Rise. FUNCTION 2022; 3:zqac040. [PMID: 38989036 PMCID: PMC11234650 DOI: 10.1093/function/zqac040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 07/12/2024] Open
Abstract
In many cell types, the rise in cytosolic Ca2+ due to opening of Ca2+ release-activated Ca2+ (CRAC) channels drives a plethora of responses, including secretion, motility, energy production, and gene expression. The amplitude and time course of the cytosolic Ca2+ rise is shaped by the rates of Ca2+ entry into and removal from the cytosol. However, an extended bulk Ca2+ rise is toxic to cells. Here, we show that the plasma membrane Ca2+ ATPase (PMCA) pump plays a major role in preventing a prolonged cytosolic Ca2+ signal following CRAC channel activation. Ca2+ entry through CRAC channels leads to a sustained sub-plasmalemmal Ca2+ rise but bulk Ca2+ is kept low by the activity of PMCA4b. Despite the low cytosolic Ca2+, membrane permeability to Ca2+ is still elevated and Ca2+ continues to enter through CRAC channels. Ca2+-dependent NFAT activation, driven by Ca2+ nanodomains near the open channels, is maintained despite the return of bulk Ca2+ to near pre-stimulation levels. Our data reveal a central role for PMCA4b in determining the pattern of a functional Ca2+ signal and in sharpening local Ca2+ gradients near CRAC channels, whilst protecting cells from a toxic Ca2+ overload.
Collapse
Affiliation(s)
- Pradeep Barak
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, UK
- Oxford Nanoimaging, Linacre House, Jordan Hill Business Park Banbury Road, Oxford OX2 8TA, UK
| | - Suneet Kaur
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| | - Erica Scappini
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| | - Charles J Tucker
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| | - Anant B Parekh
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, UK
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| |
Collapse
|
23
|
ORAI1-Regulated Gene Expression in Breast Cancer Cells: Roles for STIM1 Binding, Calcium Influx and Transcription Factor Translocation. Int J Mol Sci 2022; 23:ijms23115867. [PMID: 35682546 PMCID: PMC9180186 DOI: 10.3390/ijms23115867] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/13/2022] [Accepted: 05/20/2022] [Indexed: 12/04/2022] Open
Abstract
A remodeling of calcium homeostasis, including calcium influx via store-operated calcium entry (SOCE), is a feature of breast cancers. SOCE is critical to maintain calcium balance in the endoplasmic reticulum calcium store and is an important mechanism for calcium signaling in a variety of cell types, including breast cancer cells. The canonical mechanism of SOCE is stromal interacting molecule 1 (STIM1)-mediated activation of ORAI. Elevated ORAI1 expression is a feature of basal breast cancer cells. However, the role of ORAI1 in the regulation of transcription in breast cancer cells of the basal molecular subtype is still unclear. Using CRISPR-Cas9 gene editing, ORAI1 protein expression was disrupted in MDA-MB-231 and MDA-MB-468 basal breast cancer cells. The ORAI1 wild-type and mutants were reintroduced into ORAI1 knockout cells to study the role of ORAI1 in gene transcriptional regulation. In the absence of calcium store depletion, ORAI1 regulated PTGS2 in MDA-MB-231 cells, and this was dependent on ORAI1 pore function and STIM1 binding. The activation of SOCE by thapsigargin resulted in ORAI1-dependent increases in IL6 transcription in MDA-MB-468 cells; this was also dependent on ORAI1 pore function and STIM1 binding and was associated with the translocation of NFAT1. Given the upregulation of ORAI1 in basal breast cancer cells, our results provide further evidence that ORAI1 may contribute to cancer progression through regulation of gene expression.
Collapse
|
24
|
Synthetic gene networks recapitulate dynamic signal decoding and differential gene expression. Cell Syst 2022; 13:353-364.e6. [PMID: 35298924 DOI: 10.1016/j.cels.2022.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 11/18/2021] [Accepted: 02/17/2022] [Indexed: 12/27/2022]
Abstract
Cells live in constantly changing environments and employ dynamic signaling pathways to transduce information about the signals they encounter. However, the mechanisms by which dynamic signals are decoded into appropriate gene expression patterns remain poorly understood. Here, we devise networked optogenetic pathways that achieve dynamic signal processing functions that recapitulate cellular information processing. Exploiting light-responsive transcriptional regulators with differing response kinetics, we build a falling edge pulse detector and show that this circuit can be employed to demultiplex dynamically encoded signals. We combine this demultiplexer with dCas9-based gene networks to construct pulsatile signal filters and decoders. Applying information theory, we show that dynamic multiplexing significantly increases the information transmission capacity from signal to gene expression state. Finally, we use dynamic multiplexing for precise multidimensional regulation of a heterologous metabolic pathway. Our results elucidate design principles of dynamic information processing and provide original synthetic systems capable of decoding complex signals for biotechnological applications.
Collapse
|
25
|
Humer C, Romanin C, Höglinger C. Highlighting the Multifaceted Role of Orai1 N-Terminal- and Loop Regions for Proper CRAC Channel Functions. Cells 2022; 11:371. [PMID: 35159181 PMCID: PMC8834118 DOI: 10.3390/cells11030371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 11/16/2022] Open
Abstract
Orai1, the Ca2+-selective pore in the plasma membrane, is one of the key components of the Ca2+release-activated Ca2+ (CRAC) channel complex. Activated by the Ca2+ sensor in the endoplasmic reticulum (ER) membrane, stromal interaction molecule 1 (STIM1), via direct interaction when ER luminal Ca2+ levels recede, Orai1 helps to maintain Ca2+ homeostasis within a cell. It has already been proven that the C-terminus of Orai1 is indispensable for channel activation. However, there is strong evidence that for CRAC channels to function properly and maintain all typical hallmarks, such as selectivity and reversal potential, additional parts of Orai1 are needed. In this review, we focus on these sites apart from the C-terminus; namely, the second loop and N-terminus of Orai1 and on their multifaceted role in the functioning of CRAC channels.
Collapse
Affiliation(s)
| | | | - Carmen Höglinger
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria; (C.H.); (C.R.)
| |
Collapse
|
26
|
Bassett JJ, Robitaille M, Peters AA, Bong AHL, Taing MW, Wood IA, Sadras F, Roberts-Thomson SJ, Monteith GR. ORAI1 regulates sustained cytosolic free calcium fluctuations during breast cancer cell apoptosis and apoptotic resistance via a STIM1 independent pathway. FASEB J 2021; 36:e22108. [PMID: 34939697 DOI: 10.1096/fj.202002031rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/24/2021] [Accepted: 12/06/2021] [Indexed: 11/11/2022]
Abstract
Excessive rapid increases in cytosolic free Ca2+ have a clear association with the induction of cancer cell death. Whereas, characterizing the Ca2+ signaling events that occur during the progression of the apoptotic cascade over a period of hours or days, has not yet been possible. Now using genetically encoded Ca2+ indicators complemented with automated epifluorescence microscopy we have shown that staurosporine-induced apoptosis in MDA-MB-231 breast cancer cells was associated with delayed development of cytosolic free Ca2+ fluctuations, which were then maintained for 24 h. These cytosolic free Ca2+ fluctuations were dependent on the Ca2+ channel ORAI1. Silencing of ORAI1, but not its canonical activators STIM1 and STIM2, promoted apoptosis in this model. The pathway for this regulation implicates a mechanism previously associated with the migration of cancer cells involving ORAI1, the chaperone protein SigmaR1, and Ca2+ -activated K+ channels.
Collapse
Affiliation(s)
- John J Bassett
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Mélanie Robitaille
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Amelia A Peters
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Alice H L Bong
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Meng-Wong Taing
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Ian A Wood
- School of Mathematics and Physics, The University of Queensland, Brisbane, Queensland, Australia
| | - Francisco Sadras
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
27
|
Lu F, Li Y, Lin S, Cheng H, Yang S. Spatiotemporal regulation of store-operated calcium entry in cancer metastasis. Biochem Soc Trans 2021; 49:2581-2589. [PMID: 34854917 PMCID: PMC9436031 DOI: 10.1042/bst20210307] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 01/08/2023]
Abstract
The store-operated calcium (Ca2+) entry (SOCE) is the Ca2+ entry mechanism used by cells to replenish depleted Ca2+ store. The dysregulation of SOCE has been reported in metastatic cancer. It is believed that SOCE promotes migration and invasion by remodeling the actin cytoskeleton and cell adhesion dynamics. There is recent evidence supporting that SOCE is critical for the spatial and the temporal coding of Ca2+ signals in the cell. In this review, we critically examined the spatiotemporal control of SOCE signaling and its implication in the specificity and robustness of signaling events downstream of SOCE, with a focus on the spatiotemporal SOCE signaling during cancer cell migration, invasion and metastasis. We further discuss the limitation of our current understanding of SOCE in cancer metastasis and potential approaches to overcome such limitation.
Collapse
Affiliation(s)
- Fujian Lu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Yunzhan Li
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, United States
| | - Shengchen Lin
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, United States
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Shengyu Yang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, United States
| |
Collapse
|
28
|
Nascimento Da Conceicao V, Sun Y, Ramachandran K, Chauhan A, Raveendran A, Venkatesan M, DeKumar B, Maity S, Vishnu N, Kotsakis GA, Worley PF, Gill DL, Mishra BB, Madesh M, Singh BB. Resolving macrophage polarization through distinct Ca 2+ entry channel that maintains intracellular signaling and mitochondrial bioenergetics. iScience 2021; 24:103339. [PMID: 34816101 PMCID: PMC8591423 DOI: 10.1016/j.isci.2021.103339] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 01/21/2023] Open
Abstract
Transformation of naive macrophages into classically (M1) or alternatively (M2) activated macrophages regulates the inflammatory response. Here, we identified that distinct Ca2+ entry channels determine the IFNγ-induced M1 or IL-4-induced M2 transition. Naive or M2 macrophages exhibit a robust Ca2+ entry that was dependent on Orai1 channels, whereas the M1 phenotype showed a non-selective TRPC1 current. Blockade of Ca2+ entry suppresses pNF-κB/pJNK/STAT1 or STAT6 signaling events and consequently lowers cytokine production that is essential for M1 or M2 functions. Of importance, LPS stimulation shifted M2 cells from Orai1 toward TRPC1-mediated Ca2+ entry and TRPC1-/- mice exhibited transcriptional changes that suppress pro-inflammatory cytokines. In contrast, Orai1-/- macrophages showed a decrease in anti-inflammatory cytokines and exhibited a suppression of mitochondrial oxygen consumption rate and inhibited mitochondrial shape transition specifically in the M2 cells. Finally, alterations in TRPC1 or Orai1 expression determine macrophage polarization suggesting a distinct role of Ca2+ channels in modulating macrophage transformation.
Collapse
Affiliation(s)
| | - Yuyang Sun
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Karthik Ramachandran
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Arun Chauhan
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Amritha Raveendran
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Manigandan Venkatesan
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Bony DeKumar
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Soumya Maity
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Neelanjan Vishnu
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - George A. Kotsakis
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Paul F. Worley
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Donald L. Gill
- Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Bibhuti B. Mishra
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Muniswamy Madesh
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Brij B. Singh
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
29
|
Chaklader M, Rothermel BA. Calcineurin in the heart: New horizons for an old friend. Cell Signal 2021; 87:110134. [PMID: 34454008 PMCID: PMC8908812 DOI: 10.1016/j.cellsig.2021.110134] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/10/2021] [Accepted: 08/23/2021] [Indexed: 01/20/2023]
Abstract
Calcineurin, also known as PP2B or PPP3, is a member of the PPP family of protein phosphatases that also includes PP1 and PP2A. Together these three phosphatases carryout the majority of dephosphorylation events in the heart. Calcineurin is distinct in that it is activated by the binding of calcium/calmodulin (Ca2+/CaM) and therefore acts as a node for integrating Ca2+ signals with changes in phosphorylation, two fundamental intracellular signaling cascades. In the heart, calcineurin is primarily thought of in the context of pathological cardiac remodeling, acting through the Nuclear Factor of Activated T-cell (NFAT) family of transcription factors. However, calcineurin activity is also essential for normal heart development and homeostasis in the adult heart. Furthermore, it is clear that NFAT-driven changes in transcription are not the only relevant processes initiated by calcineurin in the setting of pathological remodeling. There is a growing appreciation for the diversity of calcineurin substrates that can impact cardiac function as well as the diversity of mechanisms for targeting calcineurin to specific sub-cellular domains in cardiomyocytes and other cardiac cell types. Here, we will review the basics of calcineurin structure, regulation, and function in the context of cardiac biology. Particular attention will be given to: the development of improved tools to identify and validate new calcineurin substrates; recent studies identifying new calcineurin isoforms with unique properties and targeting mechanisms; and the role of calcineurin in cardiac development and regeneration.
Collapse
Affiliation(s)
- Malay Chaklader
- Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Beverly A Rothermel
- Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA.
| |
Collapse
|
30
|
Oflaz FE, Koshenov Z, Hirtl M, Rost R, Bachkoenig OA, Gottschalk B, Madreiter-Sokolowski CT, Malli R, Graier WF. Near-UV Light Induced ROS Production Initiates Spatial Ca 2+ Spiking to Fire NFATc3 Translocation. Int J Mol Sci 2021; 22:8189. [PMID: 34360954 PMCID: PMC8346968 DOI: 10.3390/ijms22158189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 01/19/2023] Open
Abstract
Ca2+-dependent gene regulation controls several functions to determine the fate of the cells. Proteins of the nuclear factor of activated T-cells (NFAT) family are Ca2+ sensitive transcription factors that control the cell growth, proliferation and insulin secretion in β-cells. Translocation of NFAT proteins to the nucleus occurs in a sequence of events that starts with activating calmodulin-dependent phosphatase calcineurin in a Ca2+-dependent manner, which dephosphorylates the NFAT proteins and leads to their translocation to the nucleus. Here, we examined the role of IP3-generating agonists and near-UV light in the induction of NFATc3 migration to the nucleus in the pancreatic β-cell line INS-1. Our results show that IP3 generation yields cytosolic Ca2+ rise and NFATc3 translocation. Moreover, near-UV light exposure generates reactive oxygen species (ROS), resulting in cytosolic Ca2+ spiking via the L-type Ca2+ channel and triggers NFATc3 translocation to the nucleus. Using the mitochondria as a Ca2+ buffering tool, we showed that ROS-induced cytosolic Ca2+ spiking, not the ROS themselves, was the triggering mechanism of nuclear import of NFATc3. Collectively, this study reveals the mechanism of near-UV light induced NFATc3 migration.
Collapse
Affiliation(s)
- Furkan E. Oflaz
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Zhanat Koshenov
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Martin Hirtl
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Rene Rost
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Olaf A. Bachkoenig
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Corina T. Madreiter-Sokolowski
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Wolfgang F. Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
31
|
Kar P, Barak P, Zerio A, Lin YP, Parekh AJ, Watts VJ, Cooper DMF, Zaccolo M, Kramer H, Parekh AB. AKAP79 Orchestrates a Cyclic AMP Signalosome Adjacent to Orai1 Ca 2+ Channels. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab036. [PMID: 34458850 PMCID: PMC8394516 DOI: 10.1093/function/zqab036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/16/2021] [Accepted: 07/27/2021] [Indexed: 01/12/2023]
Abstract
To ensure specificity of response, eukaryotic cells often restrict signalling molecules to sub-cellular regions. The Ca2+ nanodomain is a spatially confined signal that arises near open Ca2+ channels. Ca2+ nanodomains near store-operated Orai1 channels stimulate the protein phosphatase calcineurin, which activates the transcription factor NFAT1, and both enzyme and target are initially attached to the plasma membrane through the scaffolding protein AKAP79. Here, we show that a cAMP signalling nexus also forms adjacent to Orai1. Protein kinase A and phosphodiesterase 4, an enzyme that rapidly breaks down cAMP, both associate with AKAP79 and realign close to Orai1 after stimulation. PCR and mass spectrometry failed to show expression of Ca2+-activated adenylyl cyclase 8 in HEK293 cells, whereas the enzyme was observed in neuronal cell lines. FRET and biochemical measurements of bulk cAMP and protein kinase A activity consistently failed to show an increase in adenylyl cyclase activity following even a large rise in cytosolic Ca2+. Furthermore, expression of AKAP79-CUTie, a cAMP FRET sensor tethered to AKAP79, did not report a rise in cAMP after stimulation, despite AKAP79 association with Orai1. Hence, HEK293 cells do not express functional active Ca2+-activated adenylyl cyclases including adenylyl cyclase 8. Our results show that two ancient second messengers are independently generated in nanodomains close to Orai1 Ca2+ channels.
Collapse
Affiliation(s)
- Pulak Kar
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Pradeep Barak
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Anna Zerio
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Yu-Ping Lin
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK,NIEHS/NIH, 111 TW Alexander Drive, Durham, NC 27709, USA
| | - Amy J Parekh
- Stoke Mandeville Hospital, Mandeville Road, Aylesbury, HP21 8AL, UK
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute of Drug Discovery, Purdue Institute of Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Dermot M F Cooper
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Holger Kramer
- Proteomics and Metabolomics Centre, Medical Research Council, London Institute of Medical Sciences, London, W12 0NN, UK
| | | |
Collapse
|
32
|
Tiffner A, Derler I. Isoform-Specific Properties of Orai Homologues in Activation, Downstream Signaling, Physiology and Pathophysiology. Int J Mol Sci 2021; 22:8020. [PMID: 34360783 PMCID: PMC8347056 DOI: 10.3390/ijms22158020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 11/21/2022] Open
Abstract
Ca2+ ion channels are critical in a variety of physiological events, including cell growth, differentiation, gene transcription and apoptosis. One such essential entry pathway for calcium into the cell is the Ca2+ release-activated Ca2+ (CRAC) channel. It consists of the Ca2+ sensing protein, stromal interaction molecule 1 (STIM1) located in the endoplasmic reticulum (ER) and a Ca2+ ion channel Orai in the plasma membrane. The Orai channel family includes three homologues Orai1, Orai2 and Orai3. While Orai1 is the "classical" Ca2+ ion channel within the CRAC channel complex and plays a universal role in the human body, there is increasing evidence that Orai2 and Orai3 are important in specific physiological and pathophysiological processes. This makes them an attractive target in drug discovery, but requires a detailed understanding of the three Orai channels and, in particular, their differences. Orai channel activation is initiated via Ca2+ store depletion, which is sensed by STIM1 proteins, and induces their conformational change and oligomerization. Upon STIM1 coupling, Orai channels activate to allow Ca2+ permeation into the cell. While this activation mechanism is comparable among the isoforms, they differ by a number of functional and structural properties due to non-conserved regions in their sequences. In this review, we summarize the knowledge as well as open questions in our current understanding of the three isoforms in terms of their structure/function relationship, downstream signaling and physiology as well as pathophysiology.
Collapse
Affiliation(s)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
33
|
Balducci V, Faris P, Balbi C, Costa A, Negri S, Rosti V, Bollini S, Moccia F. The human amniotic fluid stem cell secretome triggers intracellular Ca 2+ oscillations, NF-κB nuclear translocation and tube formation in human endothelial colony-forming cells. J Cell Mol Med 2021; 25:8074-8086. [PMID: 34288391 PMCID: PMC8358861 DOI: 10.1111/jcmm.16739] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 12/15/2022] Open
Abstract
Second trimester foetal human amniotic fluid‐derived stem cells (hAFS) have been shown to possess remarkable cardioprotective paracrine potential in different preclinical models of myocardial injury and drug‐induced cardiotoxicity. The hAFS secretome, namely the total soluble factors released by cells in their conditioned medium (hAFS‐CM), can also strongly sustain in vivo angiogenesis in a murine model of acute myocardial infarction (MI) and stimulates human endothelial colony‐forming cells (ECFCs), the only truly recognized endothelial progenitor, to form capillary‐like structures in vitro. Preliminary work demonstrated that the hypoxic hAFS secretome (hAFS‐CMHypo) triggers intracellular Ca2+ oscillations in human ECFCs, but the underlying mechanisms and the downstream Ca2+‐dependent effectors remain elusive. Herein, we found that the secretome obtained by hAFS undergoing hypoxic preconditioning induced intracellular Ca2+ oscillations by promoting extracellular Ca2+ entry through Transient Receptor Potential Vanilloid 4 (TRPV4). TRPV4‐mediated Ca2+ entry, in turn, promoted the concerted interplay between inositol‐1,4,5‐trisphosphate‐ and nicotinic acid adenine dinucleotide phosphate‐induced endogenous Ca2+ release and store‐operated Ca2+ entry (SOCE). hAFS‐CMHypo‐induced intracellular Ca2+ oscillations resulted in the nuclear translocation of the Ca2+‐sensitive transcription factor p65 NF‐κB. Finally, inhibition of either intracellular Ca2+ oscillations or NF‐κB activity prevented hAFS‐CMHypo‐induced ECFC tube formation. These data shed novel light on the molecular mechanisms whereby hAFS‐CMHypo induces angiogenesis, thus providing useful insights for future therapeutic strategies against ischaemic‐related myocardial injury.
Collapse
Affiliation(s)
- Valentina Balducci
- Department of Biology and Biotechnology "Lazzaro Spallanzani", Laboratory of General Physiology, University of Pavia, Pavia, Italy
| | - Pawan Faris
- Department of Biology and Biotechnology "Lazzaro Spallanzani", Laboratory of General Physiology, University of Pavia, Pavia, Italy
| | - Carolina Balbi
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Ambra Costa
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Sharon Negri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", Laboratory of General Physiology, University of Pavia, Pavia, Italy
| | - Vittorio Rosti
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnostic, Myelofibrosis Study Centre, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Sveva Bollini
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology "Lazzaro Spallanzani", Laboratory of General Physiology, University of Pavia, Pavia, Italy
| |
Collapse
|
34
|
Sánchez-Collado J, López JJ, Rosado JA. The Orai1-AC8 Interplay: How Breast Cancer Cells Escape from Orai1 Channel Inactivation. Cells 2021; 10:1308. [PMID: 34070268 PMCID: PMC8225208 DOI: 10.3390/cells10061308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022] Open
Abstract
The interplay between the Ca2+-sensitive adenylyl cyclase 8 (AC8) and Orai1 channels plays an important role both in the activation of the cAMP/PKA signaling and the modulation of Orai1-dependent Ca2+ signals. AC8 interacts with a N-terminal region that is exclusive to the Orai1 long variant, Orai1α. The interaction between both proteins allows the Ca2+ that enters the cell through Orai1α to activate the generation of cAMP by AC8. Subsequent PKA activation results in Orai1α inactivation by phosphorylation at serine-34, thus shaping Orai1-mediated cellular functions. In breast cancer cells, AC8 plays a relevant role supporting a variety of cancer hallmarks, including proliferation and migration. Breast cancer cells overexpress AC8, which shifts the AC8-Orai1 stoichiometry in favor of the former and leads to the impairment of PKA-dependent Orai1α inactivation. This mechanism contributes to the enhanced SOCE observed in triple-negative breast cancer cells. This review summarizes the functional interaction between AC8 and Orai1α in normal and breast cancer cells and its relevance for different cancer features.
Collapse
Affiliation(s)
| | - José J. López
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain;
| | - Juan A. Rosado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain;
| |
Collapse
|
35
|
Kar P, Lin YP, Bhardwaj R, Tucker CJ, Bird GS, Hediger MA, Monico C, Amin N, Parekh AB. The N terminus of Orai1 couples to the AKAP79 signaling complex to drive NFAT1 activation by local Ca 2+ entry. Proc Natl Acad Sci U S A 2021; 118:e2012908118. [PMID: 33941685 PMCID: PMC8126794 DOI: 10.1073/pnas.2012908118] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
To avoid conflicting and deleterious outcomes, eukaryotic cells often confine second messengers to spatially restricted subcompartments. The smallest signaling unit is the Ca2+ nanodomain, which forms when Ca2+ channels open. Ca2+ nanodomains arising from store-operated Orai1 Ca2+ channels stimulate the protein phosphatase calcineurin to activate the transcription factor nuclear factor of activated T cells (NFAT). Here, we show that NFAT1 tethered directly to the scaffolding protein AKAP79 (A-kinase anchoring protein 79) is activated by local Ca2+ entry, providing a mechanism to selectively recruit a transcription factor. We identify the region on the N terminus of Orai1 that interacts with AKAP79 and demonstrate that this site is essential for physiological excitation-transcription coupling. NMR structural analysis of the AKAP binding domain reveals a compact shape with several proline-driven turns. Orai2 and Orai3, isoforms of Orai1, lack this region and therefore are less able to engage AKAP79 and activate NFAT. A shorter, naturally occurring Orai1 protein that arises from alternative translation initiation also lacks the AKAP79-interaction site and fails to activate NFAT1. Interfering with Orai1-AKAP79 interaction suppresses cytokine production, leaving other Ca2+ channel functions intact. Our results reveal the mechanistic basis for how a subtype of a widely expressed Ca2+ channel is able to activate a vital transcription pathway and identify an approach for generation of immunosuppressant drugs.
Collapse
Affiliation(s)
- Pulak Kar
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, United Kingdom
| | - Yu-Ping Lin
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, United Kingdom
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709
| | - Rajesh Bhardwaj
- Department of Nephrology and Hypertension, University Hospital Bern, Inselspital, 3010 Bern, Switzerland
| | - Charles J Tucker
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709
| | - Gary S Bird
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709
| | - Matthias A Hediger
- Department of Nephrology and Hypertension, University Hospital Bern, Inselspital, 3010 Bern, Switzerland
| | - Carina Monico
- Micron Oxford Advanced Bioimaging Unit, Department of Biochemistry, Oxford University, Oxford OX1 3QU, United Kingdom
| | - Nader Amin
- Department of Chemistry, Oxford University, Oxford OX1 3TA, United Kingdom
| | - Anant B Parekh
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, United Kingdom;
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709
| |
Collapse
|
36
|
Thiel G, Backes TM, Guethlein LA, Rössler OG. Chromatin-embedded reporter genes: Quantification of stimulus-induced gene transcription. Gene 2021; 787:145645. [PMID: 33848575 DOI: 10.1016/j.gene.2021.145645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
Receptors and ion channels expressed on the cell surface ensure proper communication between the cells and the environment. In multicellular organism, stimulus-regulated gene transcription is the basis for communication with the environment allowing individual cells to respond to stimuli such as nutrients, chemical stressors and signaling molecules released by other cells of the organism. Hormones, cytokines, and mitogens bind to receptors and ion channels and induce intracellular signaling cascades involving second messengers, kinases, phosphatases, and changes in the concentration of particular ions. Ultimately, the signaling cascades reach the nucleus. Transcription factors are activated that respond to cellular stimulation and induce changes in gene transcription. Investigating stimulus-transcription coupling combines cell biology with genetics. In this review, we discuss the molecular biology of stimulus-induced transcriptional activators and their responsiveness to extracellular and intracellular signaling molecules and to epigenetic regulators. Stimulus-induced gene expression is measured by several methods, including detection of nuclear translocation of transcription factors, phosphorylation or DNA binding. In this article, we emphasize that the most reliable method to directly measure transcriptional activation involves the use of chromatin-embedded reporter genes.
Collapse
Affiliation(s)
- Gerald Thiel
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany.
| | - Tobias M Backes
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany
| | - Lisbeth A Guethlein
- Department of Structural Biology and Department of Microbiology & Immunology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Oliver G Rössler
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany
| |
Collapse
|
37
|
Thiel G, Schmidt T, Rössler OG. Ca 2+ Microdomains, Calcineurin and the Regulation of Gene Transcription. Cells 2021; 10:cells10040875. [PMID: 33921430 PMCID: PMC8068893 DOI: 10.3390/cells10040875] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/18/2022] Open
Abstract
Ca2+ ions function as second messengers regulating many intracellular events, including neurotransmitter release, exocytosis, muscle contraction, metabolism and gene transcription. Cells of a multicellular organism express a variety of cell-surface receptors and channels that trigger an increase of the intracellular Ca2+ concentration upon stimulation. The elevated Ca2+ concentration is not uniformly distributed within the cytoplasm but is organized in subcellular microdomains with high and low concentrations of Ca2+ at different locations in the cell. Ca2+ ions are stored and released by intracellular organelles that change the concentration and distribution of Ca2+ ions. A major function of the rise in intracellular Ca2+ is the change of the genetic expression pattern of the cell via the activation of Ca2+-responsive transcription factors. It has been proposed that Ca2+-responsive transcription factors are differently affected by a rise in cytoplasmic versus nuclear Ca2+. Moreover, it has been suggested that the mode of entry determines whether an influx of Ca2+ leads to the stimulation of gene transcription. A rise in cytoplasmic Ca2+ induces an intracellular signaling cascade, involving the activation of the Ca2+/calmodulin-dependent protein phosphatase calcineurin and various protein kinases (protein kinase C, extracellular signal-regulated protein kinase, Ca2+/calmodulin-dependent protein kinases). In this review article, we discuss the concept of gene regulation via elevated Ca2+ concentration in the cytoplasm and the nucleus, the role of Ca2+ entry and the role of enzymes as signal transducers. We give particular emphasis to the regulation of gene transcription by calcineurin, linking protein dephosphorylation with Ca2+ signaling and gene expression.
Collapse
|
38
|
Li ES, Saha MS. Optimizing Calcium Detection Methods in Animal Systems: A Sandbox for Synthetic Biology. Biomolecules 2021; 11:343. [PMID: 33668387 PMCID: PMC7996158 DOI: 10.3390/biom11030343] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 12/16/2022] Open
Abstract
Since the 1970s, the emergence and expansion of novel methods for calcium ion (Ca2+) detection have found diverse applications in vitro and in vivo across a series of model animal systems. Matched with advances in fluorescence imaging techniques, the improvements in the functional range and stability of various calcium indicators have significantly enhanced more accurate study of intracellular Ca2+ dynamics and its effects on cell signaling, growth, differentiation, and regulation. Nonetheless, the current limitations broadly presented by organic calcium dyes, genetically encoded calcium indicators, and calcium-responsive nanoparticles suggest a potential path toward more rapid optimization by taking advantage of a synthetic biology approach. This engineering-oriented discipline applies principles of modularity and standardization to redesign and interrogate endogenous biological systems. This review will elucidate how novel synthetic biology technologies constructed for eukaryotic systems can offer a promising toolkit for interfacing with calcium signaling and overcoming barriers in order to accelerate the process of Ca2+ detection optimization.
Collapse
Affiliation(s)
| | - Margaret S. Saha
- Department of Biology, College of William and Mary, Williamsburg, VA 23185, USA;
| |
Collapse
|
39
|
Uchida Y, Yamamoto Y, Sakisaka T. Trans-2-enoyl-CoA reductase limits Ca 2+ accumulation in the endoplasmic reticulum by inhibiting the Ca 2+ pump SERCA2b. J Biol Chem 2021; 296:100310. [PMID: 33482198 PMCID: PMC7949109 DOI: 10.1016/j.jbc.2021.100310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/04/2021] [Accepted: 01/14/2021] [Indexed: 12/30/2022] Open
Abstract
The endoplasmic reticulum (ER) contains various enzymes that metabolize fatty acids (FAs). Given that FAs are the components of membranes, FA metabolic enzymes might be associated with regulation of ER membrane functions. However, it remains unclear whether there is the interplay between FA metabolic enzymes and ER membrane proteins. Trans-2-enoyl-CoA reductase (TER) is an FA reductase present in the ER membrane and catalyzes the last step in the FA elongation cycle and sphingosine degradation pathway. Here we identify sarco(endo)plasmic reticulum Ca2+-ATPase 2b (SERCA2b), an ER Ca2+ pump responsible for Ca2+ accumulation in the ER, as a TER-binding protein by affinity purification from HEK293 cell lysates. We show that TER directly binds to SERCA2b by in vitro assays using recombinant proteins. Thapsigargin, a specific SERCA inhibitor, inhibits this binding. TER binds to SERCA2b through its conserved C-terminal region. TER overexpression suppresses SERCA2b ATPase activity in microsomal membranes of HEK293 cells. Depletion of TER increases Ca2+ storage in the ER and accelerates SERCA2b-dependent Ca2+ uptake to the ER after ligand-induced Ca2+ release. Moreover, depletion of TER reduces the Ca2+-dependent nuclear translocation of nuclear factor of activated T cells 4. These results demonstrate that TER is a negative regulator of SERCA2b, implying the direct linkage of FA metabolism and Ca2+ accumulation in the ER.
Collapse
Affiliation(s)
- Yasunori Uchida
- Division of Membrane Dynamics, Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
| | - Yasunori Yamamoto
- Division of Membrane Dynamics, Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
| | - Toshiaki Sakisaka
- Division of Membrane Dynamics, Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan.
| |
Collapse
|
40
|
Kutschat AP, Hamdan FH, Wang X, Wixom AQ, Najafova Z, Gibhardt CS, Kopp W, Gaedcke J, Ströbel P, Ellenrieder V, Bogeski I, Hessmann E, Johnsen SA. STIM1 Mediates Calcium-Dependent Epigenetic Reprogramming in Pancreatic Cancer. Cancer Res 2021; 81:2943-2955. [PMID: 33436389 DOI: 10.1158/0008-5472.can-20-2874] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/07/2020] [Accepted: 01/06/2021] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) displays a dismal prognosis due to late diagnosis and high chemoresistance incidence. For advanced disease stages or patients with comorbidities, treatment options are limited to gemcitabine alone or in combination with other drugs. While gemcitabine resistance has been widely attributed to the levels of one of its targets, RRM1, the molecular consequences of gemcitabine resistance in PDAC remain largely elusive. Here we sought to identify genomic, epigenomic, and transcriptomic events associated with gemcitabine resistance in PDAC and their potential clinical relevance. We found that gemcitabine-resistant cells displayed a coamplification of the adjacent RRM1 and STIM1 genes. Interestingly, RRM1, but not STIM1, was required for gemcitabine resistance, while high STIM1 levels caused an increase in cytosolic calcium concentration. Higher STIM1-dependent calcium influx led to an impaired endoplasmic reticulum stress response and a heightened nuclear factor of activated T-cell activity. Importantly, these findings were confirmed in patient and patient-derived xenograft samples. Taken together, our study uncovers previously unknown biologically relevant molecular properties of gemcitabine-resistant tumors, revealing an undescribed function of STIM1 as a rheostat directing the effects of calcium signaling and controlling epigenetic cell fate determination. It further reveals the potential benefit of targeting STIM1-controlled calcium signaling and its downstream effectors in PDAC. SIGNIFICANCE: Gemcitabine-resistant and some naïve tumors coamplify RRM1 and STIM1, which elicit gemcitabine resistance and induce a calcium signaling shift, promoting ER stress resistance and activation of NFAT signaling.
Collapse
Affiliation(s)
- Ana P Kutschat
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Feda H Hamdan
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Xin Wang
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Alexander Q Wixom
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Zeynab Najafova
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Christine S Gibhardt
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Waltraut Kopp
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Jochen Gaedcke
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Philipp Ströbel
- Department of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Elisabeth Hessmann
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Steven A Johnsen
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
41
|
Tiffner A, Maltan L, Weiß S, Derler I. The Orai Pore Opening Mechanism. Int J Mol Sci 2021; 22:E533. [PMID: 33430308 PMCID: PMC7825772 DOI: 10.3390/ijms22020533] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023] Open
Abstract
Cell survival and normal cell function require a highly coordinated and precise regulation of basal cytosolic Ca2+ concentrations. The primary source of Ca2+ entry into the cell is mediated by the Ca2+ release-activated Ca2+ (CRAC) channel. Its action is stimulated in response to internal Ca2+ store depletion. The fundamental constituents of CRAC channels are the Ca2+ sensor, stromal interaction molecule 1 (STIM1) anchored in the endoplasmic reticulum, and a highly Ca2+-selective pore-forming subunit Orai1 in the plasma membrane. The precise nature of the Orai1 pore opening is currently a topic of intensive research. This review describes how Orai1 gating checkpoints in the middle and cytosolic extended transmembrane regions act together in a concerted manner to ensure an opening-permissive Orai1 channel conformation. In this context, we highlight the effects of the currently known multitude of Orai1 mutations, which led to the identification of a series of gating checkpoints and the determination of their role in diverse steps of the Orai1 activation cascade. The synergistic action of these gating checkpoints maintains an intact pore geometry, settles STIM1 coupling, and governs pore opening. We describe the current knowledge on Orai1 channel gating mechanisms and summarize still open questions of the STIM1-Orai1 machinery.
Collapse
Affiliation(s)
- Adéla Tiffner
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (L.M.); (S.W.)
| | | | | | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (L.M.); (S.W.)
| |
Collapse
|
42
|
Bacsa B, Graziani A, Krivic D, Wiedner P, Malli R, Rauter T, Tiapko O, Groschner K. Pharmaco-Optogenetic Targeting of TRPC Activity Allows for Precise Control Over Mast Cell NFAT Signaling. Front Immunol 2021; 11:613194. [PMID: 33391284 PMCID: PMC7775509 DOI: 10.3389/fimmu.2020.613194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/17/2020] [Indexed: 11/13/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels are considered as elements of the immune cell Ca2+ handling machinery. We therefore hypothesized that TRPC photopharmacology may enable uniquely specific modulation of immune responses. Utilizing a recently established TRPC3/6/7 selective, photochromic benzimidazole agonist OptoBI-1, we set out to test this concept for mast cell NFAT signaling. RBL-2H3 mast cells were found to express TRPC3 and TRPC7 mRNA but lacked appreciable Ca2+/NFAT signaling in response to OptoBI-1 photocycling. Genetic modification of the cells by introduction of single recombinant TRPC isoforms revealed that exclusively TRPC6 expression generated OptoBI-1 sensitivity suitable for opto-chemical control of NFAT1 activity. Expression of any of three benzimidazole-sensitive TRPC isoforms (TRPC3/6/7) reconstituted plasma membrane TRPC conductances in RBL cells, and expression of TRPC6 or TRPC7 enabled light-mediated generation of temporally defined Ca2+ signaling patterns. Nonetheless, only cells overexpressing TRPC6 retained essentially low basal levels of NFAT activity and displayed rapid and efficient NFAT nuclear translocation upon OptoBI-1 photocycling. Hence, genetic modification of the mast cells' TRPC expression pattern by the introduction of TRPC6 enables highly specific opto-chemical control over Ca2+ transcription coupling in these immune cells.
Collapse
Affiliation(s)
- Bernadett Bacsa
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Annarita Graziani
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Denis Krivic
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Patrick Wiedner
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Roland Malli
- Gottfried-Schatz-Research-Center-Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Thomas Rauter
- Gottfried-Schatz-Research-Center-Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Oleksandra Tiapko
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Klaus Groschner
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| |
Collapse
|
43
|
Berlansky S, Humer C, Sallinger M, Frischauf I. More Than Just Simple Interaction between STIM and Orai Proteins: CRAC Channel Function Enabled by a Network of Interactions with Regulatory Proteins. Int J Mol Sci 2021; 22:E471. [PMID: 33466526 PMCID: PMC7796502 DOI: 10.3390/ijms22010471] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 12/27/2022] Open
Abstract
The calcium-release-activated calcium (CRAC) channel, activated by the release of Ca2+ from the endoplasmic reticulum (ER), is critical for Ca2+ homeostasis and active signal transduction in a plethora of cell types. Spurred by the long-sought decryption of the molecular nature of the CRAC channel, considerable scientific effort has been devoted to gaining insights into functional and structural mechanisms underlying this signalling cascade. Key players in CRAC channel function are the Stromal interaction molecule 1 (STIM1) and Orai1. STIM1 proteins span through the membrane of the ER, are competent in sensing luminal Ca2+ concentration, and in turn, are responsible for relaying the signal of Ca2+ store-depletion to pore-forming Orai1 proteins in the plasma membrane. A direct interaction of STIM1 and Orai1 allows for the re-entry of Ca2+ from the extracellular space. Although much is already known about the structure, function, and interaction of STIM1 and Orai1, there is growing evidence that CRAC under physiological conditions is dependent on additional proteins to function properly. Several auxiliary proteins have been shown to regulate CRAC channel activity by means of direct interactions with STIM1 and/or Orai1, promoting or hindering Ca2+ influx in a mechanistically diverse manner. Various proteins have also been identified to exert a modulatory role on the CRAC signalling cascade although inherently lacking an affinity for both STIM1 and Orai1. Apart from ubiquitously expressed representatives, a subset of such regulatory mechanisms seems to allow for a cell-type-specific control of CRAC channel function, considering the rather restricted expression patterns of the specific proteins. Given the high functional and clinical relevance of both generic and cell-type-specific interacting networks, the following review shall provide a comprehensive summary of regulators of the multilayered CRAC channel signalling cascade. It also includes proteins expressed in a narrow spectrum of cells and tissues that are often disregarded in other reviews of similar topics.
Collapse
Affiliation(s)
| | | | | | - Irene Frischauf
- Institute of Biophysics, Johannes Kepler University, 4020 Linz, Austria; (S.B.); (C.H.); (M.S.)
| |
Collapse
|
44
|
Cantonero C, Salido GM, Rosado JA, Redondo PC. PGRMC1 Inhibits Progesterone-Evoked Proliferation and Ca 2+ Entry Via STIM2 in MDA-MB-231 Cells. Int J Mol Sci 2020; 21:7641. [PMID: 33076541 PMCID: PMC7589959 DOI: 10.3390/ijms21207641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Progesterone receptor membrane component 1 (PGRMC1) has been shown to regulate some cancer hallmarks. Progesterone (P4) evokes intracellular calcium (Ca2+) changes in the triple-negative breast cancer cell lines (MDA-MB-231, MDA-MB-468, and BT-20) and in other breast cancer cell lines like the luminal MCF7 cells. PGRMC1 expression is elevated in MDA-MB-231 and MCF7 cells as compared to non-tumoral MCF10A cell line, and PGRMC1 silencing enhances P4-evoked Ca2+ mobilization. Here, we found a new P4-dependent Ca2+ mobilization pathway in MDA-MB-231 cells and other triple-negative breast cancer cells, as well as in MCF7 cells that involved Stromal interaction molecule 2 (STIM2), Calcium release-activated calcium channel protein 1 (Orai1), and Transient Receptor Potential Channel 1 (TRPC1). Stromal interaction molecule 1 (STIM1) was not involved in this novel Ca2+ pathway, as evidenced by using siRNA STIM1. PGRMC1 silencing reduced the negative effect of P4 on cell proliferation and cell death in MDA-MB-231 cells. In line with the latter observation, Nuclear Factor of Activated T-Cells 1 (NFAT1) nuclear accumulation due to P4 incubation for 48 h was enhanced in cells transfected with the small hairpin siRNA against PGRMC1 (shPGRMC1). These results provide evidence for a novel P4-evoked Ca2+ entry pathway that is downregulated by PGRMC1.
Collapse
Affiliation(s)
| | | | | | - Pedro C. Redondo
- Department of Physiology (PHYCELL Group) of Veterinary Faculty and Institute of Molecular Pathology Biomarkers (IMPB) of University of Extremadura, 10003 Caceres, Spain; (C.C.); (G.M.S.); (J.A.R.)
| |
Collapse
|
45
|
Baraniak JH, Zhou Y, Nwokonko RM, Gill DL. The Intricate Coupling Between STIM Proteins and Orai Channels. CURRENT OPINION IN PHYSIOLOGY 2020; 17:106-114. [PMID: 32954113 DOI: 10.1016/j.cophys.2020.07.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Store-operated Ca2+ entry signals are critical for cellular regulation. This intricate signaling pathway involves coupling of proteins in two different membranes: the ER-resident Ca2+-sensing STIM proteins and the highly Ca2+-selective PM Orai channels. The molecular nature of the STIM-Orai coupling interface in ER-PM junctions and consequent Orai channel gating, are processes under intense study. We describe recent developments in determining the mechanism of Orai activation through the key STIM-Orai Activating Region (SOAR) of STIM1. We describe the unexpected unimolecular coupling of STIM with Orai and explain the observed variable stoichiometry of STIM-Orai interactions. We also define the discrete C-terminal regions in Orai channels that initially latch onto STIM proteins and mediate allosteric activation of the channel. A critical "nexus" region closely associated with the STIM-activated C-terminus of Orai1, propagates the STIM-binding signal through the four tightly-associated transmembrane helices of Orai1, finally to modify the pore-forming helices and effect channel opening.
Collapse
Affiliation(s)
- James H Baraniak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Yandong Zhou
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Robert M Nwokonko
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Donald L Gill
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
46
|
Vaeth M, Kahlfuss S, Feske S. CRAC Channels and Calcium Signaling in T Cell-Mediated Immunity. Trends Immunol 2020; 41:878-901. [PMID: 32711944 DOI: 10.1016/j.it.2020.06.012] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/22/2022]
Abstract
Calcium (Ca2+) signals play fundamental roles in immune cell function. The main sources of Ca2+ influx in mammalian lymphocytes following antigen receptor stimulation are Ca2+ release-activated Ca2+ (CRAC) channels. These are formed by ORAI proteins in the plasma membrane and are activated by stromal interaction molecules (STIM) located in the endoplasmic reticulum (ER). Human loss-of-function (LOF) mutations in ORAI1 and STIM1 that abolish Ca2+ influx cause a unique disease syndrome called CRAC channelopathy that is characterized by immunodeficiency autoimmunity and non-immunological symptoms. Studies in mice lacking Stim and Orai genes have illuminated many cellular and molecular mechanisms by which these molecules control lymphocyte function. CRAC channels are required for the differentiation and function of several T lymphocyte subsets that provide immunity to infection, mediate inflammation and prevent autoimmunity. This review examines new insights into how CRAC channels control T cell-mediated immunity.
Collapse
Affiliation(s)
- Martin Vaeth
- Institute of Systems Immunology, Julius-Maximilians University of Würzburg, Würzburg, Germany; Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Sascha Kahlfuss
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
47
|
Poberezhnyi V, Marchuk O, Katilov O, Shvydiuk O, Lohvinov O. Basic concepts and physical-chemical phenomena, that have conceptual meaning for the formation of systemic clinical thinking and formalization of the knowledge of systemic structural-functional organization of the human’s organism. PAIN MEDICINE 2020. [DOI: 10.31636/pmjua.v5i2.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
From the point of view of perception and generalization processes there are complex, logic and conceptual forms of thinking. Its conceptual form is the highest result of interaction between thinking and speech. While realizing it, human uses the concept, which are logically formed thoughts, that are the meaning of representation in thinking of unity of meaningful features, relations of subjects or phenomena of objective reality. Special concepts, that are used in the science and technique are called terms. They perform a function of corresponding, special, precise marking of subjects and phenomena, their features and interactions. Scientific knowledge are in that way an objective representation of material duality in our consciousness. Certain complex of terms forms a terminological system, that lies in the basis of corresponding sphere of scientific knowledge and conditions a corresponding form and way of thinking. Clinical thinking is a conceptual form, that manifests and represents by the specialized internal speech with gnostic motivation lying in its basis. Its structural elements are corresponding definitions, terms and concepts. Cardinal features of clinical systems are consistency, criticality, justification and substantiation. Principles of perception and main concepts are represented in the article along with short descriptions of physical and chemical phenomena, that have conceptual meaning for the formation of systematic clinical thinking and formalization of systemic structural-functional organization of the human’s organism
Collapse
|
48
|
Sallinger M, Tiffner A, Schmidt T, Bonhenry D, Waldherr L, Frischauf I, Lunz V, Derler I, Schober R, Schindl R. Luminal STIM1 Mutants that Cause Tubular Aggregate Myopathy Promote Autophagic Processes. Int J Mol Sci 2020; 21:E4410. [PMID: 32575830 PMCID: PMC7352373 DOI: 10.3390/ijms21124410] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/21/2022] Open
Abstract
Stromal interaction molecule 1 (STIM1) is a ubiquitously expressed Ca2+ sensor protein that induces permeation of Orai Ca2+ channels upon endoplasmic reticulum Ca2+-store depletion. A drop in luminal Ca2+ causes partial unfolding of the N-terminal STIM1 domains and thus initial STIM1 activation. We compared the STIM1 structure upon Ca2+ depletion from our molecular dynamics (MD) simulations with a recent 2D NMR structure. Simulation- and structure-based results showed unfolding of two α-helices in the canonical and in the non-canonical EF-hand. Further, we structurally and functionally evaluated mutations in the non-canonical EF-hand that have been shown to cause tubular aggregate myopathy. We found these mutations to cause full constitutive activation of Ca2+-release-activated Ca2+ currents (ICRAC) and to promote autophagic processes. Specifically, heterologously expressed STIM1 mutations in the non-canonical EF-hand promoted translocation of the autophagy transcription factors microphthalmia-associated transcription factor (MITF) and transcription factor EB (TFEB) into the nucleus. These STIM1 mutations additionally stimulated an enhanced production of autophagosomes. In summary, mutations in STIM1 that cause structural unfolding promoted Ca2+ down-stream activation of autophagic processes.
Collapse
Affiliation(s)
- Matthias Sallinger
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Adéla Tiffner
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Tony Schmidt
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria; (T.S.); (L.W.)
| | - Daniel Bonhenry
- Center for Nanobiology and Structural Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, CZ-373 33 Nove Hrady, Czech Republic;
| | - Linda Waldherr
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria; (T.S.); (L.W.)
| | - Irene Frischauf
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Victoria Lunz
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Romana Schober
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria; (T.S.); (L.W.)
| | - Rainer Schindl
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria; (T.S.); (L.W.)
- BioTechMed-Graz, A-8010 Graz, Austria
| |
Collapse
|
49
|
Snoeck HW. Calcium regulation of stem cells. EMBO Rep 2020; 21:e50028. [PMID: 32419314 DOI: 10.15252/embr.202050028] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/14/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022] Open
Abstract
Pluripotent and post-natal, tissue-specific stem cells share functional features such as the capacity to differentiate into multiple lineages and to self-renew, and are endowed with specific cell maintenance mechanism as well as transcriptional and epigenetic signatures that determine stem cell identity and distinguish them from their progeny. Calcium is a highly versatile and ubiquitous second messenger that regulates a wide variety of cellular functions. Specific roles of calcium in stem cell niches and stem cell maintenance mechanisms are only beginning to be explored, however. In this review, I discuss stem cell-specific regulation and roles of calcium, focusing on its potential involvement in the intertwined metabolic and epigenetic regulation of stem cells.
Collapse
Affiliation(s)
- Hans-Willem Snoeck
- Columbia Center of Human Development, Columbia University Irving Medical Center, New York, NY, USA.,Division of Pulmonary Medicine, Allergy and Critical Care, Columbia University Irving Medical Center, New York, NY, USA.,Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.,Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
50
|
Graziani A, Bacsa B, Krivic D, Wiedner P, Curcic S, Schindl R, Tiapko O, Groschner K. Light-Mediated Control over TRPC3-Mediated NFAT Signaling. Cells 2020; 9:E556. [PMID: 32120825 PMCID: PMC7140526 DOI: 10.3390/cells9030556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 01/20/2023] Open
Abstract
Canonical transient receptor potential (TRPC) channels were identified as key players in maladaptive remodeling, with nuclear factor of activated T-cells (NFAT) transcription factors serving as downstream targets of TRPC-triggered Ca2+ entry in these pathological processes. Strikingly, the reconstitution of TRPC-NFAT signaling by heterologous expression yielded controversial results. Specifically, nuclear translocation of NFAT1 was found barely responsive to recombinant TRPC3, presumably based on the requirement of certain spatiotemporal signaling features. Here, we report efficient control of NFAT1 nuclear translocation in human embryonic kidney 293 (HEK293) cells by light, using a new photochromic TRPC benzimidazole activator (OptoBI-1) and a TRPC3 mutant with modified activator sensitivity. NFAT1 nuclear translocation was measured along with an all-optical protocol to record local and global Ca2+ pattern generated during light-mediated activation/deactivation cycling of TRPC3. Our results unveil the ability of wild-type TRPC3 to produce constitutive NFAT nuclear translocation. Moreover, we demonstrate that TRPC3 mutant that lacks basal activity enables spatiotemporally precise control over NFAT1 activity by photopharmacology. Our results suggest tight linkage between TRPC3 activity and NFAT1 nuclear translocation based on global cellular Ca2+ signals.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Oleksandra Tiapko
- Gottfried-Schatz-Research-Center – Biophysics; Medical University of Graz, Neue Stiftingtalstrasse 6/D04, 8010 Graz, Austria; (A.G.); (B.B.); (D.K.); (P.W.); (S.C.); (R.S.)
| | - Klaus Groschner
- Gottfried-Schatz-Research-Center – Biophysics; Medical University of Graz, Neue Stiftingtalstrasse 6/D04, 8010 Graz, Austria; (A.G.); (B.B.); (D.K.); (P.W.); (S.C.); (R.S.)
| |
Collapse
|